Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Arch Biochem Biophys ; 695: 108622, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33053365

ABSTRACT

The unloading of postural muscles leads to the changes in myosins heavy chains isoforms (MyHCs) mRNAs transcription pattern, that cause severe alterations of muscle functioning. Several transcription factors such as NFATc1 and TEAD1 upregulate slow MyHC mRNA transcription, and p38 MAP kinase can phosphorylate NFAT and TEAD1, causing their inactivation. However, the role p38 MAP kinase plays in MyHCs mRNAs transcription regulation in postural soleus muscle during unloading remains unclear. We aimed to investigate whether pharmacological inhibition of p38 MAPK during rat soleus unloading would prevent the unloading-induced slow-type MyHC mRNA transcription decrease by affecting calcineurin/NFATc1 or TEAD1 signaling. Male Wistar rats were randomly assigned to three groups: cage control (C), 3-day hindlimb suspended group (3HS) and 3-day hindlimb suspended group with the daily oral supplementation of 10 mg/kg p38 MAPK inhibitor VX-745 (3HS + VX-745). 3 days of hindlimb suspension caused the significant decreases of slow MyHC and slow-tonic myh7b mRNAs transcription as well as the decrease of NFATc1-dependent MCIP1.4 mRNA transcription in rat soleus muscles compared to the cage control. P38 MAP-kinase inhibition during hindlimb suspension completely prevented slow MyHC mRNA content decrease and partially prevented slow-tonic myh7b and MCIP1.4 mRNAs transcription decreases compared to the 3HS group. We also observed NFATc1 and TEAD1 myonuclear contents increases in the 3HS + VX-745 group compared to both 3HS and C groups (p < 0.05). Therefore, we found that p38 inhibition counteracts the unloading-induced slow MyHC mRNA transcription downregulation and leads to the activation of calcineurin/NFAT signaling cascade in unloaded rat soleus muscles.


Subject(s)
Cardiac Myosins/biosynthesis , MAP Kinase Signaling System , Muscle, Skeletal/enzymology , Myosin Heavy Chains/biosynthesis , RNA, Messenger/biosynthesis , Transcription, Genetic , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , DNA-Binding Proteins/metabolism , Male , Nuclear Proteins/metabolism , Rats , Rats, Wistar , TEA Domain Transcription Factors , Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
2.
Am J Physiol Regul Integr Comp Physiol ; 314(6): R824-R833, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29466686

ABSTRACT

Resistance training (RT) increases muscle fiber size and induces angiogenesis to maintain capillary density. Cold water immersion (CWI), a common postexercise recovery modality, may improve acute recovery, but it attenuates muscle hypertrophy compared with active recovery (ACT). It is unknown if CWI following RT alters muscle fiber type expression or angiogenesis. Twenty-one men strength trained for 12 wk, with either 10 min of CWI ( n = 11) or ACT ( n = 10) performed following each session. Vastus lateralis biopsies were collected at rest before and after training. Type IIx myofiber percent decreased ( P = 0.013) and type IIa myofiber percent increased with training ( P = 0.012), with no difference between groups. The number of capillaries per fiber increased from pretraining in the CWI group ( P = 0.004) but not the ACT group ( P = 0.955). Expression of myosin heavy chain genes ( MYH1 and MYH2), encoding type IIx and IIa fibers, respectively, decreased in the ACT group, whereas MYH7 (encoding type I fibers) increased in the ACT group versus CWI ( P = 0.004). Myosin heavy chain IIa protein increased with training ( P = 0.012) with no difference between groups. The proangiogenic vascular endothelial growth factor protein decreased posttraining in the ACT group versus CWI ( P < 0.001), whereas antiangiogenic Sprouty-related, EVH1 domain-containing protein 1 protein increased with training in both groups ( P = 0.015). Expression of microRNAs that regulate muscle fiber type (miR-208b and -499a) and angiogenesis (miR-15a, -16, and -126) increased only in the ACT group ( P < 0.05). CWI recovery after each training session altered the angiogenic and fiber type-specific response to RT through regulation at the levels of microRNA, gene, and protein expression.


Subject(s)
Cold Temperature , Immersion , Muscle Fibers, Skeletal/physiology , Neovascularization, Physiologic/physiology , Resistance Training , Capillaries/physiology , Cardiac Myosins/biosynthesis , Humans , Male , MicroRNAs/biosynthesis , Muscle Strength/physiology , Muscle, Skeletal/anatomy & histology , Muscle, Skeletal/blood supply , Muscle, Skeletal/cytology , Myosin Heavy Chains/biosynthesis , Regional Blood Flow/physiology , Vascular Endothelial Growth Factor A/biosynthesis , Young Adult
3.
J Muscle Res Cell Motil ; 38(3-4): 291-302, 2017 08.
Article in English | MEDLINE | ID: mdl-29101517

ABSTRACT

HCM, the most common inherited cardiac disease, is mainly caused by mutations in sarcomeric genes. More than a third of the patients are heterozygous for mutations in the MYH7 gene encoding for the ß-myosin heavy chain. In HCM-patients, expression of the mutant and the wildtype allele can be unequal, thus leading to fractions of mutant and wildtype mRNA and protein which deviate from 1:1. This so-called allelic imbalance was detected in whole tissue samples but also in individual cells. There is evidence that the severity of HCM not only depends on the functional effect of the mutation itself, but also on the fraction of mutant protein in the myocardial tissue. Allelic imbalance has been shown to occur in a broad range of genes. Therefore, we aimed to examine whether the MYH7-alleles are intrinsically expressed imbalanced or whether the allelic imbalance is solely associated with the disease. We compared the expression of MYH7-alleles in non-HCM donors and in HCM-patients with different MYH7-missense mutations. In the HCM-patients, we identified imbalanced as well as equal expression of both alleles. Also at the protein level, allelic imbalance was determined. Most interestingly, we also discovered allelic imbalance and balance in non-HCM donors. Our findings therefore strongly indicate that apart from mutation-specific mechanisms, also non-HCM associated allelic-mRNA expression regulation may account for the allelic imbalance of the MYH7 gene in HCM-patients. Since the relative amount of mutant mRNA and protein or the extent of allelic imbalance has been associated with the severity of HCM, individual analysis of the MYH7-allelic expression may provide valuable information for the prognosis of each patient.


Subject(s)
Alleles , Allelic Imbalance , Cardiac Myosins , Cardiomyopathy, Hypertrophic , Gene Expression Regulation, Enzymologic , Myosin Heavy Chains , Sarcomeres , Adult , Cardiac Myosins/biosynthesis , Cardiac Myosins/genetics , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/pathology , Female , Humans , Male , Middle Aged , Mutation , Myosin Heavy Chains/biosynthesis , Myosin Heavy Chains/genetics , Sarcomeres/genetics , Sarcomeres/metabolism , Sarcomeres/pathology
4.
Mol Med Rep ; 14(5): 4293-4300, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27748856

ABSTRACT

The role of myosin light chain II (MLC­II) in cellular differentiation of rat mandibular condylar chondrocytes (MCCs) induced by cyclical uniaxial compressive stress (CUCS) remains unclear. In the current study, a four­point bending system was used to apply CUCS to primary cultured MCCs from rats. It was identified that CUCS stimulated features of cellular differentiation including morphological alterations, cytoskeleton rearrangement and overproduction of proteoglycans. Furthermore, CUCS promoted runt­related transcription factor­2 (RUNX2) expression at mRNA (P<0.01) and protein levels (P<0.05) and elevated alkaline phosphatase (ALP) activity (P<0.01), which are both markers of osteogenic differentiation. Under conditions of stress, western blotting indicated that the ratio of phosphorylated MLC­II to total MLC­II was increased significantly (P<0.05). Silencing MLC­II by RNA interference reduced ALP activity (P<0.01), and eliminated RUNX2 mRNA expression (P<0.01). Addition of the MLC kinase inhibitor, ML­7, reduced the CUCS­associated upregulation of RUNX2 expression (P<0.01) and ALP activity (P<0.01). The data indicated that CUCS promoted cellular differentiation of rat primary MCCs, and this was suggested to be via the phosphorylation of MLC­II.


Subject(s)
Cardiac Myosins/genetics , Chondrocytes/cytology , Core Binding Factor Alpha 1 Subunit/biosynthesis , Mandibular Condyle/growth & development , Myosin Light Chains/genetics , Osteogenesis/genetics , Alkaline Phosphatase/antagonists & inhibitors , Alkaline Phosphatase/genetics , Animals , Azepines/administration & dosage , Cardiac Myosins/antagonists & inhibitors , Cardiac Myosins/biosynthesis , Cell Differentiation/genetics , Chondrocytes/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Cytoskeleton/genetics , Cytoskeleton/metabolism , Gene Expression Regulation, Developmental , Mandibular Condyle/cytology , Mandibular Condyle/metabolism , Myosin Light Chains/antagonists & inhibitors , Myosin Light Chains/biosynthesis , Naphthalenes/administration & dosage , Phosphorylation , Pressure , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats
5.
Am J Physiol Heart Circ Physiol ; 309(9): H1509-15, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26386113

ABSTRACT

With the advent of high-throughput DNA sequencing, the number of identified cardiomyopathy-causing mutations has increased tremendously. As the majority of these mutations affect myofilament proteins, there is a need to understand their functional consequence on contraction. Permeabilized myofilament preparations coupled with protein exchange protocols are a common method for examining into contractile mechanics. However, producing large quantities of myofilament proteins can be time consuming and requires different approaches for each protein of interest. In the present study, we describe a unified automated method to produce troponin C, troponin T, and troponin I as well as myosin light chain 2 fused to a His6-tag followed by a tobacco etch virus (TEV) protease site. TEV protease has the advantage of a relaxed P1' cleavage site specificity, allowing for no residues left after proteolysis and preservation of the native sequence of the protein of interest. After expression in Esherichia coli, cells were lysed by sonication in imidazole-containing buffer. The His6-tagged protein was then purified using a HisTrap nickel metal affinity column, and the His6-tag was removed by His6-TEV protease digestion for 4 h at 30°C. The protease was then removed using a HisTrap column, and complex assembly was performed via column-assisted sequential desalting. This mostly automated method allows for the purification of protein in 1 day and can be adapted to most soluble proteins. It has the advantage of greatly increasing yield while reducing the time and cost of purification. Therefore, production and purification of mutant proteins can be accelerated and functional data collected in a faster, less expensive manner.


Subject(s)
Cardiac Myosins/biosynthesis , Endopeptidases , Histidine , Myofibrils , Myosin Light Chains/biosynthesis , Oligopeptides , Recombinant Proteins/biosynthesis , Troponin C/biosynthesis , Troponin I/biosynthesis , Troponin T/biosynthesis , Automation , Cardiac Myosins/genetics , Chromatography, Affinity , Dextrans , Escherichia coli/genetics , Humans , Myosin Light Chains/genetics , Nickel , Recombinant Proteins/genetics , Sepharose , Troponin C/genetics , Troponin I/genetics , Troponin T/genetics
6.
Cell Transplant ; 24(12): 2479-89, 2015.
Article in English | MEDLINE | ID: mdl-25606821

ABSTRACT

In vitro expanded beating cardiac myocytes derived from induced pluripotent stem cells (iPSC-CMs) are a promising source of therapy for cardiac regeneration. Meanwhile, the cell sheet method has been shown to potentially maximize survival, functionality, and integration of the transplanted cells into the heart. It is thus hypothesized that transplanted iPSC-CMs in a cell sheet manner may contribute to functional recovery via direct mechanical effects on the myocardial infarction (MI) heart. F344/NJcl-rnu/rnu rats were left coronary artery ligated (n = 30), followed by transplantation of Dsred-labeled iPSC-CM cell sheets of murine origin over the infarct heart surface. Effects of the treatment were assessed, including in vivo molecular/cellular evaluations using a synchrotron radiation scattering technique. Ejection fraction and activation recovery interval were significantly greater from day 3 onward after iPSC-CM transplantation compared to those after sham operation. A number of transplanted iPSC-CMs were present on the heart surface expressing cardiac myosin or connexin 43 over 2 weeks, assessed by immunoconfocal microscopy, while mitochondria in the transplanted iPSC-CMs gradually showed mature structure as assessed by electron microscopy. Of note, X-ray diffraction identified 1,0 and 1,1 equatorial reflections attributable to myosin and actin-myosin lattice planes typical of organized cardiac muscle fibers within the transplanted cell sheets at 4 weeks, suggesting cyclic systolic myosin mass transfer to actin filaments in the transplanted iPSC-CMs. Transplantation of iPSC-CM cell sheets into the heart yielded functional and electrical recovery with cyclic contraction of transplanted cells in the rat MI heart, indicating that this strategy may be a promising cardiac muscle replacement therapy.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Induced Pluripotent Stem Cells/transplantation , Myocardial Contraction/physiology , Myocardial Infarction/therapy , Myocardium/cytology , Myocytes, Cardiac/cytology , Actin Cytoskeleton/metabolism , Animals , Cardiac Myosins/biosynthesis , Cell Differentiation/physiology , Cells, Cultured , Connexin 43/biosynthesis , Coronary Vessels/cytology , Female , Induced Pluripotent Stem Cells/cytology , Male , Mice , Myocardial Infarction/pathology , Rats , Rats, Inbred F344 , Stroke Volume/physiology
7.
J Biol Chem ; 290(8): 4663-4676, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25538241

ABSTRACT

Cav1.3 L-type Ca(2+) channel is known to be highly expressed in neurons and neuroendocrine cells. However, we have previously demonstrated that the Cav1.3 channel is also expressed in atria and pacemaking cells in the heart. The significance of the tissue-specific expression of the channel is underpinned by our previous demonstration of atrial fibrillation in a Cav1.3 null mutant mouse model. Indeed, a recent study has confirmed the critical roles of Cav1.3 in the human heart (Baig, S. M., Koschak, A., Lieb, A., Gebhart, M., Dafinger, C., Nürnberg, G., Ali, A., Ahmad, I., Sinnegger-Brauns, M. J., Brandt, N., Engel, J., Mangoni, M. E., Farooq, M., Khan, H. U., Nürnberg, P., Striessnig, J., and Bolz, H. J. (2011) Nat. Neurosci. 14, 77-84). These studies suggest that detailed knowledge of Cav1.3 may have broad therapeutic ramifications in the treatment of cardiac arrhythmias. Here, we tested the hypothesis that there is a functional cross-talk between the Cav1.3 channel and a small conductance Ca(2+)-activated K(+) channel (SK2), which we have documented to be highly expressed in human and mouse atrial myocytes. Specifically, we tested the hypothesis that the C terminus of Cav1.3 may translocate to the nucleus where it functions as a transcriptional factor. Here, we reported for the first time that the C terminus of Cav1.3 translocates to the nucleus where it functions as a transcriptional regulator to modulate the function of Ca(2+)-activated K(+) channels in atrial myocytes. Nuclear translocation of the C-terminal domain of Cav1.3 is directly regulated by intracellular Ca(2+). Utilizing a Cav1.3 null mutant mouse model, we demonstrate that ablation of Cav1.3 results in a decrease in the protein expression of myosin light chain 2, which interacts and increases the membrane localization of SK2 channels.


Subject(s)
Calcium Channels, L-Type/metabolism , Cell Nucleus/metabolism , Gene Expression Regulation/physiology , Myocytes, Cardiac/metabolism , Transcription, Genetic/physiology , Active Transport, Cell Nucleus/physiology , Animals , Calcium Channels, L-Type/genetics , Cardiac Myosins/biosynthesis , Cardiac Myosins/genetics , Cell Nucleus/genetics , Heart Atria/cytology , Heart Atria/metabolism , Humans , Mice , Mice, Knockout , Myocytes, Cardiac/cytology , Myosin Light Chains/biosynthesis , Myosin Light Chains/genetics , Protein Structure, Tertiary
8.
Circ Cardiovasc Genet ; 7(5): 667-76, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25077947

ABSTRACT

BACKGROUND: Nuclear reprogramming inculcates pluripotent capacity by which de novo tissue differentiation is enabled. Yet, introduction of ectopic reprogramming factors may desynchronize natural developmental schedules. This study aims to evaluate the effect of imposed transgene load on the cardiogenic competency of induced pluripotent stem (iPS) cells. METHODS AND RESULTS: Targeted inclusion and exclusion of reprogramming transgenes (c-MYC, KLF4, OCT4, and SOX2) was achieved using a drug-inducible and removable cassette according to the piggyBac transposon/transposase system. Pulsed transgene overexpression, before iPS cell differentiation, hindered cardiogenic outcomes. Delayed in counterparts with maintained integrated transgenes, transgene removal enabled proficient differentiation of iPS cells into functional cardiac tissue. Transgene-free iPS cells generated reproducible beating activity with robust expression of cardiac α-actinin, connexin 43, myosin light chain 2a, α/ß-myosin heavy chain, and troponin I. Although operational excitation-contraction coupling was demonstrable in the presence or absence of transgenes, factor-free derivatives exhibited an expedited maturing phenotype with canonical responsiveness to adrenergic stimulation. CONCLUSIONS: A disproportionate stemness load, caused by integrated transgenes, affects the cardiogenic competency of iPS cells. Offload of transgenes in engineered iPS cells ensures integrity of cardiac developmental programs, underscoring the value of nonintegrative nuclear reprogramming for derivation of competent cardiogenic regenerative biologics.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Transgenes , Actinin/biosynthesis , Animals , Cardiac Myosins/biosynthesis , Cell Differentiation , Cell Separation , Cellular Reprogramming , Connexin 43/biosynthesis , Electrophysiology , Fibroblasts/metabolism , Flow Cytometry , Genetic Techniques , Kruppel-Like Factor 4 , Mice , Microscopy, Electron , Myosin Heavy Chains/biosynthesis , Myosin Light Chains/biosynthesis , Troponin I/biosynthesis
9.
J Tissue Eng Regen Med ; 8(10): 801-10, 2014 Oct.
Article in English | MEDLINE | ID: mdl-23086759

ABSTRACT

Tissue engineering has the potential to overcome limitations associated with current management of skeletal muscle defects. This study aimed to sequentially identify a degradable phosphate glass scaffold for the restoration of muscle defects. A series of glass compositions were investigated for the potential to promote bacterial growth. Thereafter, the response of human craniofacial muscle-derived cells was determined. Glass compositions containing Fe4- and 5 mol% did not promote greater Staphylococcus aureus and Staphylococcus epidermidis growth compared to the control (p > 0.05). Following confirmation of myogenicity, further studies assessed the biocompatibility of glasses containing Fe5 mol%. Cells seeded on collagen-coated disks demonstrated comparable cellular metabolic activity to control. Upregulation of genes encoding for myogenic regulatory factors (MRFs) confirmed myofibre formation and there was expression of developmental MYH genes. The use of 3-D aligned fibre scaffolds supported unidirectional cell alignment and upregulation of MRF and developmental MYH genes. Compared to the 2-D disks, there was also expression of MYH2 and MYH7 genes, indicating further myofibre maturation on the 3-D scaffolds and confirming the importance of key biophysical cues.


Subject(s)
Absorbable Implants , Glass , Myofibrils/metabolism , Regeneration , Tissue Scaffolds , Animals , Cardiac Myosins/biosynthesis , Cells, Cultured , Humans , Myofibrils/pathology , Myosin Heavy Chains/biosynthesis , Phosphates , Rats , Staphylococcus aureus/growth & development , Staphylococcus epidermidis/growth & development , Tissue Engineering
10.
Mol Cell Biol ; 32(3): 633-51, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22144583

ABSTRACT

MicroRNA-200c (miR-200c) has been shown to suppress epithelial-mesenchymal transition (EMT), which is attributed mainly to targeting of ZEB1/ZEB2, repressors of the cell-cell contact protein E-cadherin. Here we demonstrated that modulation of miR-200c in breast cancer cells regulates cell migration, cell elongation, and transforming growth factor ß (TGF-ß)-induced stress fiber formation by impacting the reorganization of cytoskeleton that is independent of the ZEB/E-cadherin axis. We identified FHOD1 and PPM1F, direct regulators of the actin cytoskeleton, as novel targets of miR-200c. Remarkably, expression levels of FHOD1 and PPM1F were inversely correlated with the level of miR-200c in breast cancer cell lines, breast cancer patient samples, and 58 cancer cell lines of various origins. Furthermore, individual knockdown/overexpression of these target genes phenocopied the effects of miR-200c overexpression/inhibition on cell elongation, stress fiber formation, migration, and invasion. Mechanistically, targeting of FHOD1 by miR-200c resulted in decreased expression and transcriptional activity of serum response factor (SRF), mediated by interference with the translocation of the SRF coactivator mycocardin-related transcription factor A (MRTF-A). This finally led to downregulation of the expression and phosphorylation of the SRF target myosin light chain 2 (MLC2) gene, required for stress fiber formation and contractility. Thus, miR-200c impacts on metastasis by regulating several EMT-related processes, including a novel mechanism involving the direct targeting of actin-regulatory proteins.


Subject(s)
Breast Neoplasms/pathology , Fetal Proteins/metabolism , MicroRNAs/metabolism , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Breast Neoplasms/metabolism , Cardiac Myosins/biosynthesis , Cell Line, Tumor , Cell Movement , DNA-Binding Proteins/metabolism , Down-Regulation , Female , Formins , Gene Expression Regulation, Neoplastic , Humans , Myosin Light Chains/biosynthesis , Neoplasm Invasiveness , Oncogene Proteins, Fusion/metabolism , Serum Response Factor/biosynthesis , Stress Fibers/metabolism , Trans-Activators , Transforming Growth Factor beta/metabolism
11.
J Neurosci ; 31(4): 1448-60, 2011 Jan 26.
Article in English | MEDLINE | ID: mdl-21273429

ABSTRACT

Ongoing synaptic function and rapid, bidirectional plasticity are both controlled by regulatory mechanisms within dendritic spines. Spine actin dynamics maintain synapse structure and function, and cytoskeletal rearrangements in these structures trigger structural and functional plasticity. Therefore, proteins that interact with actin filaments are attractive candidates to regulate synaptic actin dynamics and, thus, synapse structure and function. Here, we have cloned the rat isoform of class II myosin heavy chain MyH7B in brain. Unexpectedly, this isoform resembles muscle-type myosin II rather than the ubiquitously expressed nonmuscle myosin II isoforms, suggesting that a rich functional diversity of myosin II motors may exist in neurons. Indeed, reducing the expression of MyH7B in mature neurons caused profound alterations to dendritic spine structure and excitatory synaptic strength. Structurally, dendritic spines had large, irregularly shaped heads that contained many filopodia-like protrusions. Neurons with reduced MyH7B expression also had impaired miniature EPSC amplitudes accompanied by a decrease in synaptic AMPA receptors, which was linked to alterations of the actin cytoskeleton. MyH7B-mediated control over spine morphology and synaptic strength was distinct from that of a nonmuscle myosin, myosin IIb. Interestingly, when myosin IIb expression and MyH7B expression were simultaneously knocked-down in neurons, a third, more pronounced phenotype emerged. Together, our data provide evidence that distinct myosin II isoforms work together to regulate synapse structure and function in cultured hippocampal neurons. Thus, myosin II motor activity is emerging as a broad regulatory mechanism for control over complex actin networks within dendritic spines.


Subject(s)
Cardiac Myosins/physiology , Myosin Heavy Chains/physiology , Neurons/metabolism , Synapses/physiology , Actins/ultrastructure , Animals , Cardiac Myosins/biosynthesis , Cardiac Myosins/genetics , Cells, Cultured , Cloning, Molecular , Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , Excitatory Postsynaptic Potentials , Female , Gene Knockdown Techniques , Hippocampus/cytology , Humans , Male , Miniature Postsynaptic Potentials , Myosin Heavy Chains/biosynthesis , Myosin Heavy Chains/genetics , Neurons/ultrastructure , Nonmuscle Myosin Type IIB/biosynthesis , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Isoforms/physiology , Protein Transport , Rats , Rats, Sprague-Dawley , Receptors, AMPA/metabolism , Synapses/ultrastructure
12.
Cell Res ; 21(4): 579-87, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21102549

ABSTRACT

Although myocyte cell transplantation studies have suggested a promising therapeutic potential for myocardial infarction, a major obstacle to the development of clinical therapies for myocardial repair is the difficulties associated with obtaining relatively homogeneous ventricular myocytes for transplantation. Human embryonic stem cells (hESCs) are a promising source of cardiomyocytes. Here we report that retinoid signaling regulates the fate specification of atrial versus ventricular myocytes during cardiac differentiation of hESCs. We found that both Noggin and the pan-retinoic acid receptor antagonist BMS-189453 (RAi) significantly increased the cardiac differentiation efficiency of hESCs. To investigate retinoid functions, we compared Noggin+RAi-treated cultures with Noggin+RA-treated cultures. Our results showed that the expression levels of the ventricular-specific gene IRX-4 were radically elevated in Noggin+RAi-treated cultures. MLC-2V, another ventricular-specific marker, was expressed in the majority of the cardiomyocytes in Noggin+RAi-treated cultures, but not in the cardiomyocytes of Noggin+RA-treated cultures. Flow cytometry analysis and electrophysiological studies indicated that with 64.7 ± 0.88% (mean ±s.e.m) cardiac differentiation efficiency, 83% of the cardiomyocytes in Noggin+RAi-treated cultures had embryonic ventricular-like action potentials (APs). With 50.7 ± 1.76% cardiac differentiation efficiency, 94% of the cardiomyocytes in Noggin+RA-treated cultures had embryonic atrial-like APs. These results were further confirmed by imaging studies that assessed the patterns and properties of the Ca(2+) sparks of the cardiomyocytes from the two cultures. These findings demonstrate that retinoid signaling specifies the atrial versus ventricular differentiation of hESCs. This study also shows that relatively homogeneous embryonic atrial- and ventricular-like myocyte populations can be efficiently derived from hESCs by specifically regulating Noggin and retinoid signals.


Subject(s)
Cell Differentiation/drug effects , Embryonic Stem Cells/metabolism , Heart Atria/metabolism , Heart Ventricles/metabolism , Myocytes, Cardiac/metabolism , Retinoids/metabolism , Signal Transduction , Action Potentials/drug effects , Base Sequence , Blotting, Western , Cardiac Myosins/biosynthesis , Cardiac Myosins/genetics , Carrier Proteins/pharmacology , Cell Line , Embryonic Stem Cells/cytology , Flow Cytometry , Fluorescent Antibody Technique , Heart Atria/embryology , Heart Ventricles/embryology , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Myocytes, Cardiac/drug effects , Myosin Light Chains/biosynthesis , Myosin Light Chains/genetics , Polymerase Chain Reaction , Retinoids/pharmacology
14.
Ann Med ; 41(5): 360-70, 2009.
Article in English | MEDLINE | ID: mdl-19165643

ABSTRACT

AIM: The differentiation efficiencies of human embryonic stem cell (hESC) lines differ from each other. To assess this in more detail we studied the cardiac differentiation of eight hESC lines derived in the same laboratory. RESULTS: Substantial variation in growth and in the ability to form beating areas was seen between the different hESC lines; line HS346 gave the best efficiency (9.4%), while HS293 did not differentiate into beating colonies at all. Nine germ layer and differentiation markers were quantified during early differentiation in four hESC lines. The expression levels of Brachyury T, MESP1 and NKX2.5 were highest in the most efficient cardiac line (HS346). A systematic characterization of the beating cells revealed proper cardiac marker expression, electrophysiological activity, and pharmacological response. CONCLUSIONS: The hESC lines derived in the same laboratory varied considerably in their potential to differentiate into beating cardiomyocytes. None of the expression markers could clearly predict cardiac differentiation potential, although the expression of early cardiomyogenic genes was upregulated in the best cardiac line. The proper cardiomyocyte characteristics and pharmacological response indicate that these cells could be used as a model for human cardiomyocytes in pharmacological and toxicological analyses when investigating new heart medications or cardiac side-effects.


Subject(s)
Cardiac Myosins/genetics , Cell Differentiation/physiology , Embryonic Stem Cells/ultrastructure , Gene Expression Regulation, Developmental , Myocytes, Cardiac/ultrastructure , RNA/genetics , Biomarkers/metabolism , Calcium Channel Blockers/pharmacology , Cardiac Myosins/biosynthesis , Cell Line , Electrophysiologic Techniques, Cardiac , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Germ Layers/drug effects , Germ Layers/metabolism , Germ Layers/ultrastructure , Humans , Immunohistochemistry , Membrane Potentials/drug effects , Microscopy, Immunoelectron , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Verapamil/pharmacology
15.
Stem Cells Dev ; 17(6): 1131-40, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19006452

ABSTRACT

Previous studies have suggested that mesenchymal stem cells (MSCs) can differentiate into smooth muscle-like cells. However their functionalities remain questionable. The aim of this study was to investigate the functionality of MSCs differentiated into smooth muscle (SM) in vitro by SM-inducing medium. MSCs have been isolated from rat bone marrow and cultured in SM-inducing medium. After 21 days in culture, messenger RNA and specific SM proteins such as myosin heavy chain and myosin light chain 2 were expressed in the in vitro differentiated MSCs to a similar level of that in freshly isolated SM cells (SMCs). At the electrophysiological level, MSCs presented an outward K+ current with an IK(DR) component and IK(Ca) component. In vitro differentiation induced an enhancement of the IK(Ca) current to a level similar to that observed in aortic SMCs. Calcium homeostasis measurements revealed that both differentiated and undifferentiated MSCs responded to extracellular adenosine triphosphate (ATP) in a similar fashion to SMCs. However MSCs failed to contract in response to ATP. This data shows that despite specific SM protein expression and modification of electrophysiological properties similar to that of aortic SMCs, MSCs cultured in differentiation medium failed to display contractile properties. These results underline the necessity to find the ideal cultured conditions to induce complete SMC function.


Subject(s)
Cell Differentiation/physiology , Membrane Potentials/physiology , Mesenchymal Stem Cells/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Adenosine Triphosphate/pharmacology , Animals , Calcium/metabolism , Cardiac Myosins/biosynthesis , Cell Differentiation/drug effects , Cells, Cultured , Homeostasis/physiology , Membrane Potentials/drug effects , Mesenchymal Stem Cells/cytology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Myosin Heavy Chains/biosynthesis , Myosin Light Chains/biosynthesis , Rats , Time Factors
16.
Gene Expr Patterns ; 7(7): 738-45, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17638597

ABSTRACT

Mrf4 (Myf6) is a member of the basic helix-loop-helix (bHLH) myogenic regulatory transcription factor (MRF) family, which also contains Myod, Myf5 and myogenin. Mrf4 is implicated in commitment of amniote cells to skeletal myogenesis and is also abundantly expressed in many adult muscle fibres. The specific role of Mrf4 is unclear both because mrf4 null mice are viable, suggesting redundancy with other MRFs, and because of genetic interactions at the complex mrf4/myf5 locus. We report the cloning and expression of an mrf4 gene from zebrafish, Danio rerio, which shows conservation of linkage to myf5. Mrf4 mRNA accumulates in a subset of terminally differentiated muscle fibres in parallel with myosin protein in the trunk and fin. Although most, possibly all, trunk muscle expresses mrf4, the level of mRNA is dynamically regulated. No expression is detected in muscle precursor cell populations prior to myosin accumulation. Moreover, mrf4 expression is not detected in head muscles, at least at early stages. As fish mature, mrf4 expression is pronounced in the region of slow muscle fibres.


Subject(s)
Myogenic Regulatory Factors/biosynthesis , Myogenic Regulatory Factors/physiology , Amino Acid Sequence , Amnion/cytology , Animals , Cardiac Myosins/biosynthesis , Cardiac Myosins/physiology , Gene Expression Regulation , Models, Biological , Molecular Sequence Data , Muscles/metabolism , Myogenic Regulatory Factor 5/metabolism , Myogenin/metabolism , Myosin Light Chains/biosynthesis , Myosin Light Chains/physiology , Phylogeny , Sequence Homology, Amino Acid , Tissue Distribution , Zebrafish
17.
Biochem Biophys Res Commun ; 354(4): 999-1003, 2007 Mar 23.
Article in English | MEDLINE | ID: mdl-17276389

ABSTRACT

Cardiomyocyte regeneration is limited in adult life and is not sufficient to compensate for cell loss with myocardial infarction. Hence, the identification of a useful source of cardiomyocyte progenitors is of great interest for possible use in regenerative therapy. In this study, we isolated stem cells derived from human subcutaneous adipose tissue. The expression of Nkx2.5 and GATA-4 can be observed by PCR directly after extraction and during cultivation in some of these cells. Cardiac Troponin T and myosin light chain-2v become positive after 12 days of cultivation. To define respective factors responsible for spontaneous differentiation, we measured VEGF level in ADSC conditioned medium. Our data showed that ADSC secrete significant amount of VEGF (283.5pg per microgram DNA) and that anti-VEGF receptor antibodies blocked the cardiac differentiation. In conclusion, we demonstrated the spontaneous differentiation of human subcutaneous adipose-derived stem cells into a cardiomyocyte phenotype under standard culturing conditions.


Subject(s)
Cell Differentiation/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Stem Cells/physiology , Vascular Endothelial Growth Factor A/physiology , Animals , Cardiac Myosins/biosynthesis , Cells, Cultured , GATA4 Transcription Factor/biosynthesis , Homeodomain Proteins/biosynthesis , Humans , Myosin Light Chains/biosynthesis , Rats , Subcutaneous Fat/cytology , Transcription Factors/biosynthesis , Troponin T/biosynthesis
18.
Circ Res ; 100(3): 354-62, 2007 Feb 16.
Article in English | MEDLINE | ID: mdl-17234970

ABSTRACT

The sinoatrial node, which resides at the junction of the right atrium and the superior caval vein, contains specialized myocardial cells that initiate the heart beat. Despite this fundamental role in heart function, the embryonic origin and mechanisms of localized formation of the sinoatrial node have not been defined. Here we show that subsequent to the formation of the Nkx2-5-positive heart tube, cells bordering the inflow tract of the heart tube give rise to the Nkx2-5-negative myocardial cells of the sinoatrial node and the sinus horns. Using genetic models, we show that as the myocardium of the heart tube matures, Nkx2-5 suppresses pacemaker channel gene Hcn4 and T-box transcription factor gene Tbx3, thereby enforcing a progressive confinement of their expression to the forming Nkx2-5-negative sinoatrial node and sinus horns. Thus, Nkx2-5 is essential for establishing a gene expression border between the atrium and sinoatrial node. Tbx3 was found to suppress chamber differentiation, providing an additional mechanism by which the Tbx3-positive sinoatrial node is shielded from differentiating into atrial myocardium. Pitx2c-deficient fetuses form sinoatrial nodes with indistinguishable molecular signatures at both the right and left sinuatrial junction, indicating that Pitx2c functions within the left/right pathway to suppress a default program for sinuatrial node formation on the left. Our molecular pathway provides a mechanism for how pacemaker activity becomes progressively relegated to the most recently added components of the venous pole of the heart and, ultimately, to the junction of the right atrium and superior caval vein.


Subject(s)
Body Patterning/physiology , Gene Expression Regulation, Developmental/physiology , Heart Atria/embryology , Heart Ventricles/embryology , Homeodomain Proteins/physiology , Ion Channels/biosynthesis , Sinoatrial Node/embryology , T-Box Domain Proteins/physiology , Transcription Factors/physiology , Animals , Atrial Natriuretic Factor , Biomarkers , Body Patterning/genetics , Cardiac Myosins/biosynthesis , Cardiac Myosins/genetics , Connexins/biosynthesis , Connexins/genetics , Cyclic Nucleotide-Gated Cation Channels , Gene Expression Regulation, Developmental/genetics , Genes, Reporter , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Imaging, Three-Dimensional , In Situ Hybridization , Ion Channels/genetics , Mice , Mice, Knockout , Mice, Transgenic , Myocardium/metabolism , Myosin Light Chains/biosynthesis , Myosin Light Chains/genetics , Natriuretic Peptide, C-Type/biosynthesis , Natriuretic Peptide, C-Type/genetics , Protein Precursors/biosynthesis , Protein Precursors/genetics , Recombinant Fusion Proteins/physiology , Sinoatrial Node/cytology , T-Box Domain Proteins/biosynthesis , T-Box Domain Proteins/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Troponin I/biosynthesis , Troponin I/genetics , Gap Junction alpha-5 Protein , Homeobox Protein PITX2
19.
Acta Pharmacol Sin ; 27(3): 311-20, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16490167

ABSTRACT

AIM: To investigate effects of icariin on cardiac gene expression and the modulation of nitric oxide (NO) signal transduction during the differentiation of embryonic stem (ES) cells into cardiomyocytes in vitro. METHODS: The expression levels of cardiac developmental-dependent genes were measured using reverse transcription-polymerase chain reaction (RT-PCR). The chronotropic responses of cardiomyocytes to b-adrenoceptor stimulation were determined. The levels of cAMP and cGMP in ES cells were measured using radioimmunoassay. Endogenous NO levels were measured by using the Griess reaction. Aminoguanidine (AG) was used to confirm the influence of icariin on the endogenous NO signal pathway. RESULTS: Icariin significantly elevated mRNA levels of cardiac transcription factors GATA4 and Nkx2.5, and cardiac-specific alpha-MHC, MLC-2v and beta-AR genes in a concentration- and time-dependent manner (P<0.05). Cardiomyocytes derived from embryoid body (EB) treated with icariin were more sensitive to isoprenaline (P<0.01). Treatment of ES cells with icariin resulted in a continued elevation in the cAMP/cGMP ratio before a shift to the cardiomyocyte phenotype (P<0.05). AG decreased the NO level, and delayed and decreased the incidence of contracting EB to only approximately 35% on d 5+11, an effect that could be rescued by icariin. When cells were cocultured with icariin and AG, the percentage of beating EB reached a peak level of 73% on d 5+11 (P<0.05). CONCLUSION: The inducible effects of icariin were partly related to increase in the expression of cardiac developmental-dependent genes, and elevation of the cAMP/cGMP ratio in ES cells, as well as upregulation of endogenous NO generation during the early stages of cardiac development.


Subject(s)
Cell Differentiation/drug effects , Flavonoids/pharmacology , Myocytes, Cardiac/cytology , Nitric Oxide/metabolism , Stem Cells/cytology , Animals , Cardiac Myosins/biosynthesis , Cardiac Myosins/genetics , Cell Line , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Drugs, Chinese Herbal/isolation & purification , Drugs, Chinese Herbal/pharmacology , Embryo, Mammalian , Epimedium/chemistry , Flavonoids/isolation & purification , GATA4 Transcription Factor/biosynthesis , GATA4 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Mice , Myocytes, Cardiac/metabolism , Myosin Light Chains/biosynthesis , Myosin Light Chains/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Adrenergic, beta/biosynthesis , Receptors, Adrenergic, beta/genetics , Signal Transduction , Transcription Factors/biosynthesis , Transcription Factors/genetics
20.
Circulation ; 112(9 Suppl): I123-8, 2005 Aug 30.
Article in English | MEDLINE | ID: mdl-16159803

ABSTRACT

BACKGROUND: Angiogenesis is enhanced after transplantation of vascular endothelial growth factor (VEGF)-expressing cells into a myocardial scar. Insulin-like growth factor I (IGF-I) may induce hypertrophy and inhibit apoptosis. We evaluated the effect of cell-based IGF-I and VEGF multigene therapy on left ventricular (LV) function, cell survival, and apoptosis after bone marrow cell (BMC) transplantation. METHODS AND RESULTS: Female Lewis rats underwent left anterior descending ligation 3 weeks before transplantation with male donor BMC, BMC transfected with VEGF (BMC+VEGF), IGF-I (BMC+IGF-I), VEGF and IGF-I (BMC+VEGF+IGF-I), or medium without cells (control) (n=4 per group x 5 groups x 4 time points). Three days and 1, 2, and 4 weeks after transplantation, VEGF and IGF-I expression was quantitated by real-time polymerase chain reaction, cell survival by polymerase chain reaction for sry2, apoptosis by TUNEL staining, LV function by echocardiography and myosin heavy chain, and light chain and troponin I by Western blot. One week after transplantation, IGF-I expression in the scar and border zone was greatest in BMC+IGF-I and BMC+VEGF+IGF-I rats (P<0.05). VEGF expression in the scar and border zone was greatest in BMC+VEGF and BMC+VEGF+IGF-I hearts (P<0.05). Transplanted cell survival was lowest in BMC, intermediate in BMC+VEGF and BMC+IGF-I, and greatest in BMC+VEGF+IGF-I (P<0.05). Apoptotic indices were significantly reduced in BMC+VEGF+IGF-I, BMC+VEGF, and BMC+IGF-I (P<0.05). Two and 4 weeks after transplantation, LV ejection fraction was lowest in control, intermediate in BMC, BMC+VEGF, and BMC+IGF-I, and greatest in BMC+VEGF+IGF-I (P<0.05). CONCLUSIONS: Transplantation of VEGF- and IGF-I-expressing BMC reduced apoptosis, maximized transplanted cell survival, and enhanced LV function. Multimodal cell-based gene therapy may maximize the benefits of cell transplantation.


Subject(s)
Bone Marrow Transplantation , Genetic Therapy , Insulin-Like Growth Factor I/physiology , Mesenchymal Stem Cell Transplantation , Myocardial Infarction/therapy , Vascular Endothelial Growth Factor A/physiology , Ventricular Dysfunction, Left/therapy , Ventricular Function, Left , Animals , Apoptosis , Bone Marrow Cells/cytology , Cardiac Myosins/biosynthesis , Cardiac Myosins/genetics , Cell Survival , Cells, Cultured/transplantation , Cicatrix/pathology , Female , Graft Survival , Insulin-Like Growth Factor I/biosynthesis , Insulin-Like Growth Factor I/genetics , Male , Myocardial Infarction/complications , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myosin Heavy Chains/biosynthesis , Myosin Heavy Chains/genetics , Myosin Light Chains/biosynthesis , Myosin Light Chains/genetics , Neovascularization, Physiologic , Rats , Rats, Inbred Lew , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/physiology , Stroke Volume , Transfection , Troponin I/biosynthesis , Troponin I/genetics , Ultrasonography , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...